Sie sind auf Seite 1von 11

Cellular Markers of Muscle Atrophy in

Chronic Obstructive Pulmonary Disease


Pamela J. Plant1, Dina Brooks2, Marie Faughnan1, Tanya Bayley1, James Bain3, Lianne Singer4, Judy Correa1,
Dawn Pearce5, Matthew Binnie1, and Jane Batt1
1
Department of Medicine and 5Department of Radiology and St. Michaels Hospital, University of Toronto, Toronto, Ontario, Canada;
2
Department of Physical Therapy, University of Toronto and Toronto Rehab Institute, Toronto, Ontario, Canada; 3Department of Surgery,
McMaster University, Hamilton, Ontario, Canada; and 4Department of Medicine, Toronto General Hospital, University Health Network,
University of Toronto, Toronto, Ontario, Canada

Skeletal muscle atrophy in individuals with advanced chronic ob- be enhanced by decreased regenerative capacity of the muscle
structive pulmonary disease (COPD) is associated with diminished (reviewed in References 68). The molecular mechanisms un-
quality of life, increased health resource use, and worsened survival. derlying skeletal muscle atrophy have recently begun to be
Muscle wasting results from an imbalance between protein degra- elucidated through in vitro experimentation in myotube cultures
dation and synthesis, and is enhanced by decreased regenerative and in rodent models of disease.
repair. We investigated the activation of cellular signaling networks Although muscle proteolysis is mediated via the coordinated
known to mediate muscle atrophy and regulate muscle regenerative
efforts of several cellular networks, including, for example,
capacity in rodent models, in individuals with COPD (FEV1 , 50%
oxidative stress responses, activation of calpains, and non-
predicted). Nine patients with COPD and nine control individuals
were studied. Quadriceps femoris muscle isometric contractile force
lysosomal proteases, it is the ubiquitinproteasome pathway
and cross-sectional area were confirmed to be significantly smaller in that predominates (6, 812). The ubiquitin ligases, atrogin-1,
the patients with COPD compared with control subjects. The vastus muscle-specific RING finger protein (MuRF)1, and neural
lateralis muscle was biopsied and muscle transcript and/or protein precursor cellexpressed developmentally down-regulated
levels of key components of ubiquitin-mediated proteolytic systems (Nedd)4, key enzymes regulating ubiquitin-mediated protein
(MuRF1, atrogin-1, Nedd4), inflammatory mediators (IkBa, NF- degradation, are variably up-regulated in multiple rodent
kBp65/p50), AKT network (AKT, GSK3b, p70S6 kinase), medi- models of skeletal muscle atrophy, ranging from traumatic
ators of autophagy (beclin-1, LC3), and myogenesis (myogenin, denervation to chronic metabolic diseases, such as cancer and
MyoD, Myf5, myostatin) were determined. Atrogin-1 and Nedd4, diabetes (1317). More recently, activation of the autophagic/
two ligases regulating ubiquitin-mediated protein degradation and lysosomal system has also been recognized to contribute to
myostatin, a negative regulator of muscle growth, were significantly muscle atrophy and, at least in vitro, autophagy seems to play
increased in the muscle of patients with COPD. MuRF1, Myf5, a substantial role in the loss of muscle protein (18, 19).
myogenin, and MyoD were not differentially expressed. There were
In muscle atrophy associated with generalized inflammatory
no differences in the level of phosphorylation of AKT, GSK3b,
states (20, 21), the inflammatory cytokine TNF-a causes muscle
p70S6kinase, or IkBa, activation of NF-kBp65 or NF-kBp50, or level
of expression of beclin-1 or LC3, suggesting that AKT signaling was
proteolysis and inhibits muscle regenerative capacity by in-
not down-regulated and the NF-kB inflammatory pathway and ducing oxidative stress, and destabilizing MyoD (20, 22)
autophagy were not activated in the COPD muscle. We conclude through NF-kB activation. MyoD is a muscle transcription fac-
that muscle atrophy associated with COPD results from the re- tor essential to myogenesis and muscle regeneration. NF-kB
cruitment of specific regulators of ubiquitin-mediated proteolytic activation also mediates expression of the ubiquitin ligase,
pathways and inhibition of muscle growth. MuRF1, thus enhancing proteasome-mediated protein degra-
dation (23, 24).
Keywords: vastus lateralis; ubiquitin ligase; myostatin; neural precursor It is also known that the engagement of muscle atrophy
cellexpressed developmentally down-regulated 4; atrogin-1 signaling networks results in the concurrent down-regulation of
the signaling pathways that induce muscle hypertrophy. Acti-
Skeletal muscle atrophy is a critical phenomenon that occurs in
vation of the AKT (protein kinase B) pathway, and signaling
individuals with chronic obstructive pulmonary disease
through its downstream targets, such as glycogen synthase
(COPD). It is associated with diminished exercise capacity (1)
and quality of life (2), increased health resource use and health kinase (GSK)-3b and p70S6 kinase (70-kD ribosomal protein
care costs (3), and is a powerful negative predictor of survival S6 kinase), results in muscle hypertrophy (8, 25). This pathway
(4, 5). Loss of skeletal muscle mass results from decreased is down-regulated/deactivated in models of muscle wasting,
muscle protein synthesis and increased proteolysis, and this may demonstrating a key finding that there is a reciprocal link
between the muscle hypertrophic and atrophic signaling net-
works (8, 25, 26).
The muscle regulatory transcription factors, myogenin and
(Received in original form October 8, 2008 and in final form May 8, 2009) myogenic factor (Myf)5, which induce myoblast differentiation
This work was supported by Canadian Institutes of Health Research New and muscle regeneration and myostatin, a negative regulator of
Investigator Awards (D.B. and J.B.), a Keenan Foundation Fellowship, St Michaels muscle growth, can also influence muscle mass, as demonstrated
Hospital (P.P.), and by an Ontario Thoracic Society Grant-in-Aid Award ( J.B., D.B., in genetic rodent models (reviewed in References 7, 27).
and M.F.). Few studies have assessed cellular signaling of muscle
Correspondence and requests for reprints should be addressed to Jane Batt, atrophy in humans, and thus the relevance of these networks
M.D., Ph.D., Room 7344, Medical Sciences Building, 1 Kings College Circle, and effectors of atrophy in human disease is not clear. In
University of Toronto, Toronto, ON, M5S 1A8 Canada. E-mail: jane.batt@
addition, in the myriad of signaling networks activated to induce
utoronto.ca
muscle atrophy in rodent models, there is some specificity of
Am J Respir Cell Mol Biol Vol 42. pp 461471, 2010
Originally Published in Press as DOI: 10.1165/rcmb.2008-0382OC on June 11, 2009 pathway recruitment to the atrophy model assessed. Given this
Internet address: www.atsjournals.org variability, we need to determine which cellular signaling
462 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 42 2010

networks are active in contributing to the clinically significant bar, as previously described (34, 35). Isometric contraction is reflective
muscle atrophy of COPD. This will provide future targets for of the amount of time a muscle group can perform and exercise.
pharmacologic manipulation, with the goal of improving quality
of life, functional status, and, potentially, survival in COPD. Vastus Lateralis Percutaneous Biopsy
Thus, in this study, we sought to identify the cellular signaling Biopsy of the vastus lateralis muscle was performed under local
pathways activated in atrophied skeletal muscle of patients with anesthetic using a modified Bergstrom needle, as previously described
severe COPD by measuring mRNA transcript and/or protein (36). Briefly, under sterile conditions, the skin and subcutaneous tissue
expression levels of key signaling network components in were anesthetized, and a small incision was made in the outer fascial
layer with a scalpel blade. The Bergstrom needle was advanced through
biopsies of the quadriceps muscles.
the incision roughly 1 cm into the muscle, suction was applied as the
trochar was advanced, and several pieces of muscle tissue were obtained.
The needle was withdrawn under counter pressure, the skin closed with
MATERIALS AND METHODS
a single suture, and a pressure dressing applied. Extracted tissue was
Patient and Control Populations snap frozen in liquid nitrogen for RNA and protein extraction.
Patients with COPD were recruited from respirology clinics at
St. Michaels Hospital (SMH) and the Toronto General Hospital, RNA Extraction and Real-Time RT-PCR
University of Toronto. Patients were deemed eligible if they had Real-time RT PCR was used to assess expression levels of transcripts
a physician diagnosis of COPD, as defined by the Canadian Thoracic for the ubiquitin ligases, atrogin-1, MuRF1, and the myogenic regula-
Society guidelines (28), and severe disease (FEV1 , 50% predicted). tory factors (MRFs), myogenin, Myf5, and myostatin (an inhibitor of
Exclusion criteria included: (1) the use of systemic glucocorticoid muscle growth), and mediators of autophagy, beclin-1 and autophagy-
therapy in the preceding 90 days or the chronic use of medication that related protein LC (LC)-3. Transcript levels were assessed as opposed
causes muscle atrophy; (2) comorbidities associated with muscle to protein levels, because good commercial antibodies were not avail-
atrophy (infection, diabetes, end-stage renal disease on dialysis, active able and/or the limited amount of protein available necessitated that
cancer); (4) COPD exacerbation or hospitalization in the previous mRNA levels be determined. These genes do not produce proteins that
3 months; (5) HIV, hepatitis B, or hepatitis C infection; (6) age greater require phosphorylation for activation, and determination of the level
than 75 years; and (8) institutionalized care or required assistance for of expression of the transcripts was therefore deemed to be adequate.
activities of daily living. Tissue snap-frozen in liquid nitrogen was homogenized in Trizol
A healthy age-matched control population was drawn from volun- (1 ml/mg tissue; Life Technologies, Burlington, ON, Canada) to isolate
teer participants registered with the SMH Healthy Lung Database and total muscle RNA, per the manufacturers recommendations. RNA
through advertisements that were placed throughout the hospitals. quality was assessed with agarose gel electrophoresis and the Agilent
These individuals had no known lung disease and did not fulfill any of 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA), and quanti-
the study exclusion criteria. The study protocol was approved by the fied by absorption spectrophotometry at 260 and 280 nm.
research ethics boards at Toronto General Hospital and SMH. All To remove any contaminating genomic DNA, the muscle total
participating subjects provided informed consent. RNA samples were treated with DNA-free (Ambion, Austin, TX), per
the manufacturers instructions. cDNA was then generated from the
Function and Quality of Life Measurements total RNA from each patient and control subject in a first-strand
synthesis reaction using Superscript II Reverse Transcriptase (Invitro-
Pulmonary disease severity and degree of impairment and disability gen, Burlington, ON, Canada) at 428C for 2 hours, according to the
were characterized using pulmonary function testing, BODE score, and manufacturers instructions. Relative quantitative real-time RT-PCR
6-minute walk distance as outlined briefly here. was subsequently performed with an Applied Biosystems 7900 system
Pulmonary function testing. Spirometry was performed in and SYBR Green Master Mix (Applied Biosystems, Foster City, CA)
accordance with recommended techniques detailed by the American for atrogin-1, MuRF1, myogenin, Myf5, myostatin, beclin-1, and
Thoracic Society guidelines (29). The primary parameters of assess- microtubule-associated protein 1A/1B-light chain 3 (LC3). Primers
ment were FEV1, FVC, and the FEV1:FVC ratio. were designed using Primer Express software (Applied Biosystems)
The 6-minute walk test. The 6-minute walk test was used to (Table 1).
assess functional exercise capacity, according to American Thoracic For each gene, real-time PCR was performed in triplicate wells on
Society established criteria (30, 31). We applied standardized instruc- cDNA generated from the reverse transcription of 10 ng of total RNA.
tion and encouragement. At baseline, the patients and control subjects Negative controls for each gene included no template (water) and no
completed at least two training walks to account for learning effect. reverse transcriptase (10 ng RNA). The PCR amplification consisted of
BODE score. The BODE score incorporates body mass index 10-minute denaturation, followed by 40 cycles of amplification (15 s at
(BMI), FEV1% predicted, score on the modified Medical Research 958C, 60 s at 608C). After amplification, amplicons were melted and the
Council dyspnea scale, and 6-minute walk distance to estimate survival resulting dissociation curve assessed to ensure a single product.
in COPD (32). The BODE score was calculated for all participants. Aliquots (10 ml) of all products were run on DNA polyacrylamide
gels to ensure the presence of a single amplicon of the expected size.
Assessment of Body Composition, Muscle Mass, and Strength The real-time experiments were repeated twice for all genes and all
subjects. The expression level of each transcript for each individual
Muscle mass and body composition were determined using clinical (patient or control subject) was determined relative to a pooled RNA
measures (BMI, sum of five skin folds, circumferential waist girth), and reference generated in the laboratory, using the relative quantification
the midthigh quadriceps femoris muscle cross-sectional area (CSA) (DDCT) technique, according to Applied Biosystems instructions. Two
was determined as previously described (33). Briefly, computed housekeeping genestyrosine 3 monooxygenase/tryptophan 5 mon-
tomography imaging of the right thigh, halfway between the pubic oxygenase (YWHAZ), or hydroxymethylbilane synthetase (HMBS)
symphysis and the inferior condyle of the femur, using a Light Speed were used. Primer amplification efficiencies were equal for all genes
QXi 4 slice helical scanner (General Electric, Milwaukee, WI), was tested and both housekeeping genes (data not shown).
performed with the subject in the supine position. Each image was 10
to 20 mm thick, and the muscle identified as tissue with a density of 40
to 100 Hounsfield units. Images were analyzed and the CSA of muscle Protein Extraction and Western Blotting
determined by a single investigator, blinded as to the categorization of Proteins were extracted by homogenizing the frozen muscle in lysis
each test subject. buffer (5 mM Tris-HCL [pH 8.0], 1 mM EDTA, 1 mM EGTA, 1 mM
Muscle strength was determined using isometric contractile strength b-mercaptoethanol, 1% glycerol with 1 mM PMSF, 10 mg/ml leupeptin,
testing of the quadriceps femoris muscle. Isometric contraction of the 10 mg/ml aprotinin, 1 mM orthovanadate) with a Polytron PTE 1200E
quadriceps femoris was measured with the use of a strain gauge, with the homogenizer (Kinematica, Lucerne, Switzerland) in three 30-second
subject seated on a chair extending the knee against a stationary metal pulses, and homogenates were cleared by centrifuging at 1,600 3 g for
Plant, Brooks, Faughnan, et al.: Markers of Muscle Atrophy in COPD 463

TABLE 1. REAL-TIME RT-PCR PRIMER SEQUENCES


Gene Forward Primer Reverse Primer

HMBS tgc aac ggc gga aga aaa agc tgg ctc ttg cgg gta c
YWHAZ gca atg atg tac tgt ctc ttt tgg aa taa cgg tag taa tct cct ttc att ttc a
Atrogin-1 gca cgt gct cag cga aga atc tgc cgc tcg gag aag t
MuRF1 tcc agc aga cac tga acc aga a tcc att ttg cac caa tgt aga aa
Myf5 agg tca acc agg ctt tcg aa gat gta gcg gat ggc att cc
Myostatin ttg aga ccc gtc gag act cct a ttc aga gat cgg att cca gta tac c
Myogenin gct gta tga gac atc ccc cta ctt cgt agc ctg gtg gtt cga a
Beclin-1 agg aac tca cag ctc cat tac aat ggc tcc tct cct gag tt
LC3 atg tca aca tga gcg agt tgg t ctg gtt cac cag cag gaa gaa

Definition of abbreviations: HMBS, hydroxymethylbilane synthetase; LC3, microtubule-associated protein 1A/1B-light chain 3;
MuRF, muscle-specific RING finger protein; Myf, myogenic factor; YWHAZ, tyrosine 3 monooxygenase/tryptophan 5 monoxygenase.

10 minutes at 48C. The supernatant (soluble fraction) was centrifuged RESULTS


further for 10 minutes at 48C and 10,000 3 g. The pellet (insoluble
fraction) was washed with PBS, resuspended, dissolved by sonication in Characterization of the COPD Patient and
200 ml buffer containing protease/phosphatase inhibitors (as described Control Populations
previously here), and further centrifuged (10,000 3 g, 48C) for
10 minutes, and the supernatant designated as the insoluble fraction. A total of 94 individuals with severe COPD were assessed for
Protein lysate (7.5 mg) was separated by SDS-PAGE, immunoblotted, study inclusion. Of these, 37 were not eligible based on
and bands detected using chemiluminescence generated by Super- exclusion criteria. Of the remaining 57 eligible patients, 9
Signal West Femto Chemiluminescent Substrate (Thermo Scientific, provided informed consent and took part in the study. Nine
Waltham, MA) acquired with a charge-coupled device camera (Bio- age-matched healthy control individuals provided informed
Rad VersaDoc, Hercules, CA), and quantified using Quantity One consent and took part in the study.
software (Bio-Rad). Primary antibodies used included: actin (1:200 To characterize our study populations, body composition,
dilution; Sigma-Aldrich, St. Louis MO); AKT; phospho-AKT (Ser pulmonary function, and functional capacity were assessed and
473); phospho-GSK3a/b (Ser 21/9); inhibitory kB (IkB)-a; phospho
data are provided in Table 2. As expected, individuals with
IkB-a (Ser 32/36); NF-kB p65; phospho-NF-kB p65 (Ser 536); p70 S6
kinase (1:1,000 dilution; all from Cell Signaling, Boston, MA); lamin
severe COPD had significantly lower 6-minute walk distances
A 1 C and MuRF1 (1:1,000 dilution; both from Abcam, Cambridge, and significantly higher BODE scores compared with control
MA); phosphop70S6 kinase (Thr 389, 1:1,000 dilution; Cell Signaling); individuals. There were no differences between the COPD and
MyoD (1:500 dilution; Santa Cruz, Santa Cruz, CA); GAPDH control populations with respect to BMI, percent body fat, or
(1:100,000 dilution; Abcam); GSK3a/b (1:10,000 dilution; Invitrogen, sum of five skin folds. The distribution of males and females was
Carlsbad, CA); and a-tubulin (1:50,000 dilution; Sigma). Secondary not equal between groups, with more females in the control
antibodies consisted of horseradish peroxidase (HRP)conjugated anti- group. Two male patients with COPD were involved in a pre
mouse, anti-rabbit, and anti-goat at 1:20,000 dilution. Phosphorylated lung transplant rehabilitation program, and one female patient
proteins were normalized in expression to their total unphosphorylated undertook regular aerobic and resistance training at least three
forms. MyoD, Nedd4, and MuRF1 were normalized to two house-
times per week. The remaining patients with COPD were all
keeping proteins, a-tubulin and GAPDH. Given the limited sample
available from the muscle biopsies, the levels of some proteins of fully independent and intermittently exercised. Four control
interest were not determined in all study participants. Total numbers participants (two male and two female) undertook regular
assessed are detailed in the results section. aerobic training greater than three times per week. The re-
mainder intermittently exercised.
Activated NF-kB(p65) and NF-kB(p50) Assays To confirm the presence of muscle atrophy in the patients
Quantitation of the expression of active NF-kB(p65) and active NF- with COPD, we measured CSA of the quadriceps femoris
kB(p50) was completed using the Thermo Scientific Transcription muscle. Patients with COPD demonstrated smaller quadriceps
Factor Kits for NF-kB(p65) and NF-kB(p50) (nos. 89858 and 89859; CSA (61.26 6 5.36 cm2; n 5 9) compared with control in-
ThermoFisher Scientific, Waltham, MA), as directed by the manufac- dividuals (89.88 6 4.59 cm2; n 5 9) as determined by computed
turer. Briefly, 7 mg of protein lysates were incubated with the NF-kB tomography imaging (P , 0.05; Figure 1A). Similarly, the
binding biotinylated consensus sequence DNA (p65 or p50) adherent strength of the quadriceps was lower in the patients with
to the streptavidin-coated 96-well plates provided. Only the active NF- COPD than the control subjects (11.95 6 1.06 kg; n 5 8 versus
kB (p65 or p50) binds to the consensus sequence, and is subsequently
detected with antiNF-kB(p65) or NF-kB(p50) primary antibody, an
HRP-conjugated secondary antibody and a chemiluminescent sub- TABLE 2. SUBJECT CHARACTERISTICS
strate (all provided in the kits). Chemiluminescence was detected in
Patient Subjects Control Subjects
an EnVision Multilabel Plate Reader Model 2102 (Perkin Elmer, Characteristics (n 5 9) (n 5 9) Significance
Waltham, MA) and reported as relative chemiluminescence units.
Specificity of the binding of the activated transcription factors to the Age, yr 64.0 6 2.1 59.6 6 1.3 N.S.
consensus sequence DNA was ensured by using control wild-type and Sex, male/female 5/4 3/6
mutant NF-kB (p65 or p50) competitor duplexes supplied with the kit. Body mass index 24.4 6 1.1 25.9 6 1.8 N.S.
Body fat, % 26.8 6 2.0 26.8 6 2.0 N.S.
Statistical Analysis Sum of five skin folds, cm 58.7 6 7.5 65.3 6 5.0 N.S.
Waist circumference, cm 92.7 6 3.0 99.8 6 11.9 N.S.
Continuous data are reported as mean and SE, and were compared using FEV1, L/s 0.88 6 0.09 2.87 6 0.22 P , 0.05
unpaired Students t test or Wilcoxons rank-sum test after testing for FEV1% predicated 35.1 6 2.5 115.1 6 7.9 P , 0.05
normal distribution (Shapiro-Wilk). When comparing multiple indepen- BODE 4.78 6 0.45 0.11 6 0.11 P , 0.05
dent means, a one-way ANOVA was first performed to confirm a 6-min walk, m 393 6 32 617 6 22 P , 0.05
difference across all groups prior to comparison of individuals means.
Results were considered significant if P values were less than 0.05. Definition of abbreviations: N.S., not significant.
464 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 42 2010

Figure 1. Quadriceps cross-sectional area (CSA)


and power. Patients with chronic obstructive
pulmonary disease (COPD) demonstrate signif-
icantly decreased CSA of the quadriceps femoris
muscle (A) and are weaker (B) than the age-
matched control subjects. Significant differ-
ences in muscle area between patients and
control subjects are maintained even when data
are assessed in sex-specific groups (C). Quadri-
ceps of female patients are significantly weaker
than in female control subjects (D), and male
patients are significantly weaker than male
control subjects, but male patients are not
significantly weaker than female control sub-
jects. F, female; M, male.

23.46 6 3.02 kg, n 5 9; P , 0.05; Figure 1B). One male patient fraction between patients with COPD and control individuals
declined strength testing. (data not shown).
Due to the uneven distribution of males and females in
the patient and control populations, we assessed differences Mediators of Muscle Regenerative CapacityMuscle
on a sex-specific basis. Female patients muscle area (46.37 6 Regulatory Transcription Factors and Myostatin
5.95 cm2; n 5 4) was smaller than female control individuals
Real-time RT-PCR was performed for myostatin, a negative
(81.59 6 2.92 cm2; n 5 6; P , 0.05; Figure 1C). Male patients
regulator of muscle growth and the muscle regulatory transcrip-
muscle area (73.18 6 1.55 cm2; n 5 5) was significantly smaller
tion factors, myogenin and Myf5, which induce myoblast
than both male control subjects (106.50 6 1.94 cm2; n 5 3; P ,
differentiation and muscle regeneration. Myostatin transcript
0.05) and female control subjects (P , 0.05; Figure 1C). Sex-
levels (Figure 3A) were significantly higher in the patients with
specific comparisons were also performed for strength (Figure
COPD (3.1 6 0.8fold change; n 5 9) compared with the
1D). Female patients quadriceps strength was significantly
control group (0.9 6 0.1fold change; n 5 9; P , 0.05). Neither
weaker than female control subjects (11.4 6 1.9 kg [n 5 4]
Myf5 (Figure 3B) nor myogenin (Figure 3C) demonstrated
versus 20.1 6 2.6 kg [n 5 6]; P , 0.05). Male patients were
significant differences in expression between patients with
weaker than male control subjects (12.5 6 1.1 kg [n 5 4] versus
COPD (n 5 9) and the control population (n 5 9) (Myf5,
30.1 6 6.5 kg [n 5 3]; P , 0.05). Male patients, however, were
0.96 6 0.18 versus 0.88 6 0.15fold change [P . 0.05], and
not significantly weaker than female control subjects (P 5 0.062).
myogenin, 1.32 6 0.27 versus 0.96 6 0.16fold change [P .
Ubiquitin LigasesMediators of Protein Degradation by 0.05]). Similarly, there was no difference in the level of
the Proteasome expression of the proregenerative MyoD in the soluble fraction
of muscle protein lysates in patients with COPD (0.14 6 0.05;
Good-quality RNA was extracted for all study participants. n 5 8) versus control individuals (0.06 6 0.03; n 5 8; P . 0.05;
Patients with COPD demonstrated a significant increase in the Figures 3D and 3E).
level of atrogin-1 transcript expression (2.8 6 0.5fold change;
n 5 9) compared with the control individuals (1.4 6 0.2fold
change; n 5 9; P , 0.05) (Figure 2A). There was no difference NF-kB Signaling NetworkInflammatory mediators
in the expression level of MuRF1 transcripts between patients Phosphorylation of IkBa leads to its proteolytic degradation,
with COPD (10.7 6 2.1fold change; n 5 9) and control and subsequent release and activation of NF-kB transcription
individuals (8.8 6 2.2fold change; n 5 9; P . 0.05) (Figure factors. Total IkBa and phosphorylated IkBa (pIkBa) protein
2B), nor was there any difference in MuRF1 proteins levels in levels (Figure 4) were determined in the soluble fraction of
the soluble fraction of muscle protein lysates between patients muscle protein lysates by Western blotting. There was no
with COPD (0.22 6 0.10; n 5 9) and control individuals (0.11 6 significant difference in the level of phosphorylation of IkBa
0.03; n 5 9; P . 0.05) (Figures 2D and 2F). in the patients with COPD compared with the control popula-
In contrast, Nedd4 was increased in expression in the tion (Figure 4), as determined by measuring the ratio of
insoluble fraction of muscle protein lysates (insoluble myofi- phosphorylated IkBa/total IkBa (patients with COPD, 0.34 6
brils) in the patients with COPD (0.37 6 0.08; n 5 9) versus 0.11 [n 5 9], versus control subjects, 0.22 6 0.09 [n 5 8]; P .
control subjects (0.09 6 0.03; n 5 9; P , 0.05) (Figures 2C and 0.05) or when normalizing phosphorylated IkBa levels to
2E). Nedd4 was also detectable in the soluble fraction of protein GAPDH (patients with COPD, 0.18 6 0.05 [n 5 9] versus
lysates, but no differences in expression were seen in this control subjects, 0.20 6 0.04 [n 5 8]; P . 0.05).
Plant, Brooks, Faughnan, et al.: Markers of Muscle Atrophy in COPD 465

Figure 2. Ubiquitin ligases. Atrogin-1 mRNA levels (A) were increased in patients with chronic obstructive pulmonary disease (COPD) versus control
individuals, as determined by relative quantification with real-time RT-PCR. There was no difference in muscle-specific RING finger protein (MuRF) 1
transcript levels (B) between patients and control subjects. mRNA levels in patients and control subjects were compared with a common reference
standard. Tyrosine 3 monooxygenase/tryptophan 5 monoxygenase (YWHAZ) served as the housekeeping gene (HKG), and similar data were
obtained using hydroxymethylbilane synthetase (HMBS) as the HKG (data not shown). Neural precursor cellexpressed developmentally down-
regulated (Nedd) 4 protein levels (C ) were increased in the insoluble fraction of patient muscle protein lysates compared with control subjects, as
determined by SDS PAGE, Western blotting, and charge-coupled device camera image acquisition, and quantification of the chemiluminescent
signal. Nedd4 expression was normalized to a-tubulin, and a representative Western blot is shown (E ). Similar results were obtained normalizing
Nedd4 to GAPDH (data not shown). There was no difference in MuRF1 protein (D) expression in the soluble fraction of muscle protein lysates
between patients and control subjects. MuRF1 protein levels were normalized to a-tubulin, and a representative Western blot is shown (F ). Similar
results were obtained normalizing to GAPDH (data not shown). Co, control individual; N.S., not significant; Pt, patient; P . 0.05.

Quantitation of activation of NF-kB(p65) and NF-kB(p50) AKT (Figure 6) in the patients with COPD (0.44 6 0.13; n 5 8)
was determined by measuring the chemiluminescence signal versus the control population (0.30 6 0.10; n 5 9) (P . 0.05), the
generated by binding of the activated transcription factors to level of phosphorylation of GSK3b (Figure 7) in the patients with
their respective DNA consensus sequences fixed in a 96-well COPD (0.58 6 0.16, n 5 8) versus the control population (0.74 6
plate and detected with antiNF-kB(p65) and antiNF-kB(p50) 0.17; n 5 9) (P . 0.05) or level of phosphorylation of p70S6
primary antibodies and HRP-linked secondary antibodies. There kinase (Figure 8) in the patients with COPD (0.34 6 0.10; n 5 8)
was no significant difference in the level of expression of active versus the control population (0.29 6 0.08; n 5 9) (P . 0.05).
NF-kB(p65) in patients with COPD (7.23 6 1.6 3 105 chem-
iluminescence units; n 5 6) versus control subjects (6.4 6 1.7 3 Mediators of AutophagyBeclin-1 and LC3
105 chemiluminescence units; n 5 6) (P . 0.05; Figure 5A). There was no difference in the level of expression of beclin-1
Similarly, there was no difference in the level of expression of transcripts in patients with COPD (6.7 6 1.2 fold change; n 5 9)
active NF-kB(p50) between patients with COPD (1.9 6 0.8 3 compared with control individuals (7.4 6 1.1 fold change; n 5 9)
106 chemiluminescence units; n 5 5) and control subjects (1.2 6 (P . 0.05; Figure 9A). Similarly, there was no difference in the
0.5 3 106 chemiluminescence units; n 5 6) (P . 0.05; Figure 5B). level of expression of LC3 transcripts between patients with
COPD (52.4 6 16.5 fold change; n 5 9) and control individuals
AKT Signaling NetworkMediators of Muscle Hypertrophy (54.3 6 8.5 fold change; n 5 9) (P . 0.05; Figure 9B).
Total AKT and phosphorylated AKT, total GSK3b and phos-
phorylated GSK3b, and total p70S6 kinase and phosphorylated DISCUSSION
p70S6 kinase protein levels were determined in the soluble
fraction of the muscle protein lysates by Western blot analysis. Skeletal muscle atrophy and weakness occur in individuals with
There were no differences in the level of phosphorylation of advanced COPD, and this reduction in muscle mass is associ-
466 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 42 2010

Figure 3. Muscle-regulatory transcription


factors and myostatin. Myostatin mRNA
levels (A) were significantly increased in
patients with chronic obstructive pulmonary
disease (COPD) compared with control sub-
jects, as determined by relative quantifica-
tion with real-time RT-PCR. There were no
differences in the level of expression of (B)
myogenic factor (Myf)5 transcripts or (C)
myogenin transcripts between patients with
COPD and control subjects. Transcript levels
in patients and control subjects were com-
pared with a common reference standard.
YWHAZ served as the HKG. Results were
similar using HMBS as the HKG (data not
shown). MyoD (D) protein levels in the
soluble fraction of muscle protein lysates
were not significantly different between
patients with COPD and control subjects,
as determined by SDS PAGE, Western blot-
ting, and charge-coupled device camera
image acquisition, and quantification of
the chemiluminescent signal. MyoD expres-
sion was normalized to a-tubulin. (E) A
representative Western blot is shown. Simi-
lar results were obtained normalizing MyoD
to GAPDH (data not shown). Co, control;
N.S., not significant; Pt, patient; P . 0.05.

ated with increased morbidity, mortality, health resource use, atrophy (reviewed in References 68) have been derived from
and cost (1, 35). The significant advances made in the past experimentation conducted largely in rodent models of disease,
decade delineating the cellular signaling pathways and key and the clinical relevance of these findings is not known. In
mediators responsible for the development of skeletal muscle the present study, we sought to identify the cellular signaling

Figure 4. Inhibitory kB (IkB)-a.


(A) There was no significant
difference in the level of IkBa
phosphorylation in the pa-
tients with chronic obstructive
pulmonary disease (COPD)
versus control individuals. To-
tal and phosphorylated IkBa
(IkBa and pIkBa) protein
levels were determined in the
soluble cytoplasmic fraction of
muscle protein lysates by SDS-
PAGE, Western blotting, and
charge-coupled device camera
image acquisition, and quan-
tification of the chemilumines-
cent signal. pIkBa expression
was normalized to total IkBa
and to GAPDH. (B) A repre-
sentative Western blot is
shown. Positive (Pos) and neg-
ative (Neg) controls for IkB
phosphorylation consisted of
protein lysates from HeLa cell
cultures treated with TNF-a
(20 ng/ml 3 5 min) and un-
treated, respectively. Co, con-
trol; N.S., not significant; Pt,
patient; P . 0.05.
Plant, Brooks, Faughnan, et al.: Markers of Muscle Atrophy in COPD 467

Figure 6. AKT. (A) There was no difference in the level of phosphory-


lation of AKT in the patients with chronic obstructive pulmonary
disease (COPD) versus the control population. Total AKT and phos-
phorylated AKT (pAKT) protein levels were determined by SDS-PAGE,
Western blotting, charge-coupled device camera image acquisition,
and quantitation of the chemiluminescent signal in the soluble fraction
of muscle protein lysates. pAKT protein levels were normalized to total
Figure 5. NF-kB(p65) and NF-kB(p50) activation. Expression levels of AKT protein levels. (B) A representative Western blot is shown. Protein
the active forms of NF-kB(p65) and NF-kB(p50) proteins were de- lysates of 293T cell cultures treated with insulin, and untreated, served
termined in muscle protein lysates using the ThermoScientific Transcrip- as positive (Pos) and negative (Neg) controls, respectively, for AKT
tion Factor Kits for NF-kB(p65) and NF-kB(p50). Activated p65 and p50 phosphorylation. Western blotting for GAPDH served as a loading
bound to the NF-kBbinding consensus sequence DNA immobilized in control. Co, control; N.S., not significant; Pt, patient; P . 0.05.
96-well plates was detected using antiNF-kB(p65) and NF-kB(p50)
antibodies, horseradish peroxidaselinked secondary antibodies and
recently, a third ligase, Nedd4, has also been recognized (13,
a chemiluminescent substrate. The chemiluminescent signal was quan-
tified in an EnVision multilabel plate reader. There was no significant
16, 39) to contribute to this process in rodent models.
difference in the level of expression of active NF-kB(p65) (A) or The up-regulation of atrogin-1 and Nedd4 noted in our
NF-kB(p50) (B) in the patients with chronic obstructive pulmonary patients with COPD infers an increase in ubiquitinproteasome
disease (COPD) versus control individuals. N.S., not significant; P . mediated proteolysis in the COPD muscle. Interestingly we did
0.05 (59, 60). not find MuRF1 to be increased. Discordance in the up-
regulation of these three ubiquitin ligases has been previously
reported, with the pattern and temporality of ligase recruitment
networks responsible for the development of muscle atrophy in dependent upon the etiology of the muscle atrophy. Ottenheijm
individuals with COPD. Our patients with COPD were care- and colleagues (40) reported enhanced proteasomal degrada-
fully selected to exclude comorbid illness and medical therapies tion in the diaphragms of patients with COPD, and found
that could affect muscle mass. Therefore, the significant atrophy increased transcript levels of atrogin-1, but not MuRF1, in the
and weakness of the quadriceps femoris muscle demonstrated diaphragmatic muscle. Nedd4 expression was not investigated
most likely results from the advanced but stable COPD in our in this population. Nedd4 is increased in atrophy associated with
patient population. We report the novel observations of up- denervation or unloading (8, 13, 41), but is not increased in
regulation of myostatin and the ubiquitin ligase, Nedd4, and muscle atrophy resulting from diabetes, renal failure, or star-
confirm up-regulation of a second ubiquitin ligase, atrogin-1 vation in mice or rats (16, 17, 39). Atrophy resulting from
(which has been previously reported [37]), in the atrophied denervation in rodents involves the up-regulation of atrogin-1,
vastus lateralis muscle of individuals with severe COPD. MuRF1, and Nedd4, but atrogin-1 and MuRF1 increases are
Skeletal muscle protein loss is mediated by a series of short lived, with persistent increases seen only in the level of
intracellular signaling networks, but ubiquitin-dependent pro- Nedd4 (13). Thus, there is specificity of these ubiquitin ligases
teolytic mechanisms are the predominant mechanisms respon- to the systemic process responsible for the development of
sible for the development of muscle atrophy (reviewed in atrophy and in the temporality of its progression. This is not
References 6, 812). Protein ubiquitination is a highly ordered surprising, as the target substrates identified to date for each
process whereby proteins to be degraded are tagged with enzyme differ (13, 16, 4244). Clearly, in the human population,
multiple ubiquitin moieties, which serve as recognition markers, further study is necessary to fully understand the implications of
targeting proteins for subsequent proteolytic cleavage by the this specificity in muscle atrophy arising from different disease
26S proteasome or lysosome. Ubiquitin ligases are the critical processes. Our novel finding that Nedd4 is up-regulated, and
enzymes that both link ubiquitin moieties to the protein confirmation of atrogin-1 up-regulation in the COPD patient
targeted for degradation and confer specificity to the system population, is significant, as it provides possible targets for
by interacting directly with the target protein through well- future therapeutic intervention in the treatment of COPD.
defined proteinprotein interaction domains (38). The ubiquitin It is interesting that we note an increase in Nedd4 in the
ligases, atrogin-1 and MuRF1, are key regulators of muscle muscle of patients with COPD, whereas up-regulation of Nedd4
atrophy in multiple rodent models (14, 15, 17), and, more in rodent models of atrophy has been limited to unloaded or
468 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 42 2010

Figure 7. Glycogen synthase kinase (GSK)3b. (A) There was


no significant difference in the level of phosphorylation of
GSK3b in the patients with chronic obstructive pulmonary
disease (COPD) versus the control population. Total GSK3b
and phosphorylated GSK3b (pGSK3b) protein levels in the
soluble fractions of muscle protein lysates were determined
by SDS-PAGE, Western blotting, charge-coupled device cam-
era image acquisition, and quantification of the chemilumi-
nescent signal. pGSK3b protein levels were normalized to
total GSK3b levels. (B) A representative Western blot is
shown. Protein lysates of 293T cell cultures treated with
insulin, and untreated, served as positive (Pos) and negative
(Neg) controls, respectively, for GSK3b phosphorylation.
Western blotting for GAPDH served as a loading control.
Co, control; N.S., not significant; Pt, patient; P . 0.05.

denervated muscle. The unloading model in rats (hindlimb deletion of the myostatin gene demonstrate a hypermuscular
suspension) is an extreme muscle disuse model. It may be that phenotype (27). Increased myostatin expression in both serum
there is a graded effect of muscle loading on Nedd4 expression, and muscle was associated with decreased muscle mass in HIV-
with relative muscle disuse resulting in only slightly increased positive patients with muscle wasting (49).
Nedd4 expression. This would not have been noted in the rat Our finding of increased levels of myostatin transcripts in the
models of diabetes, renal failure, or starvation, because these patients with COPD implies that the loss of muscle mass in our
animals retain full activity. Our patients with COPD were COPD population is attributed to impaired muscle growth, in
ambulatory; their muscles are loaded and innervated, but they addition to active muscle degradation, reiterating the notion
may experience relative disuse compared with control individ- that atrophy is a multifactorial process rather than the result of
uals, resulting in increased Nedd4 expression. An alternative a single biochemical pathway/signal. We cannot comment on
explanation is that we noted increased Nedd4 expression only in the signaling pathways engaged downstream of myostatin, as we
the insoluble myofibrillar fraction of protein lysates, not in the did not interrogate the cell cycle regulators. However, as we did
soluble fraction. Koncarevic and colleagues (16) localized not demonstrate any changes in the levels of MyoD, myogenin,
Nedd4 to the sarcolemma in healthy rodent muscle, and
assessed levels only in cytoplasmic and crude membrane
fractions. We have noted increased expression of Nedd4 in
the insoluble myofibrillar fraction of protein lysates of rodent
muscle atrophying from denervation (39). In other organs and
cells, Nedd4 is expressed throughout the cytoplasm, with
membrane localization occurring only upon target substrate
binding (45, 46). Given the varying subcellular distribution, and
the fact that the downstream target substrates of Nedd4 in
muscle remain largely undefined, it is not possible to state in
which fraction increased Nedd4 expression is necessary for the
development of muscle atrophy.
In addition to enhanced protein breakdown, alterations in
the regeneration of muscle can influence muscle mass. Muscle
satellite cells are the population of precursor cells that re-
generate muscle and maintain muscle mass (47, 48). These cells
are normally quiescent, but they are activated to proliferate
after muscle injury, terminally differentiating to mature myo-
cytes, and fusing to form new myofibers or repair damaged
myofibers. The MRFs, MyoD, Myf5, and myogenin, are essen-
tial to the processes of muscle self-renewal, and specify satellite
Figure 8. p70S6 Kinase. (A) There was no significant difference in the
cell terminal differentiation into muscle cells. We found no
level of phosphorylation of p70S6 kinase in the soluble fraction of
differences in the level of expression of Myf5, myogenin, or
muscle protein lysates of patients with chronic obstructive pulmonary
MyoD between patients and control subjects, suggesting that
disease (COPD) versus the control population. Total p70S6 kinase and
myoblast differentiation in COPD muscles is unaffected. phosphorylated p70S6 kinase (p-p70S6 kinase) protein levels were
Myostatin is a member of the transforming growth factor b determined by SDS-PAGE, Western blotting, charge-coupled device
family, and is a negative regulator of skeletal muscle develop- camera image acquisition, and quantification of the chemiluminescent
ment and growth. Myostatin inhibits proliferation of satellite signal. p-p70S6kinase expression was normalized to total p70S6 kinase
cells/myoblasts by up-regulating expression of p21 and decreas- expression. (B) A representative Western blot is shown. Protein lysates
ing Cdk2 and phosphorylated Rb, which results in cell cycle of 293T cell cultures treated with insulin, and untreated, served as
withdrawal (27). Myostatin also appears to inhibit satellite cell positive (Pos) and negative (Neg) controls, respectively, for p70S6
differentiation via the down-regulation of various MRFs, in- kinase phosphorylation. Western blotting for GAPDH served as a load-
cluding MyoD, Myf5, and myogenin. Mice with a targeted ing control. Co, control; N.S., not significant; Pt, patient; P . 0.05.
Plant, Brooks, Faughnan, et al.: Markers of Muscle Atrophy in COPD 469

Other investigations have recently addressed the question


of the presence of a proinflammatory milieu in the skeletal
muscle of stable patients with COPD compared with healthy
control subjects. Neither Barreiro and colleagues (56) nor Crul
and colleagues (57) were able to demonstrate local inflamma-
tion in the vastus lateralis muscle of clinically and weight-
stable patients with COPD by assessing the level of expression
of various cytokines. Barreiro did, however, demonstrate
increased oxidative stress in the COPD muscle. Thus, studies
performed in humans to date, including our results here,
suggest that inflammatory mediators and signaling via NF-
kB(p65) are not active in the muscle of clinically stable
Figure 9. Autophagy factors beclin-1 and microtubule-associated pro- patients with COPD, and do not contribute to the develop-
tein 1A/1B-light chain 3 (LC3). There was no difference in the level of ment of muscle atrophy in those with normal weight and BMI.
expression of beclin-1 or LC3 transcripts between patients with chronic This does not preclude the possibility that, during acute illness,
obstructive pulmonary disease (COPD) and control individuals, as this pathway may be recruited, but our study did not address
determined by relative quantification with real-time RT-PCR. mRNA this issue.
levels in patients and control subjects were compared with a common Recently, NF-kB(p50) has been shown to mediate muscle
reference standard. YWHAZ served as the HKG, and similar data were atrophy associated with unloading, independent of activation of
obtained using HMBS as HKG (data not shown). N.S., not significant; p65 (58). The downstream substrate targets of p50 that mediate
P . 0.05. this effect are unknown, and, to date, no other upstream
stimulants that engage this atrophic pathway have been identi-
or Myf5, it is probable that myostatin is affecting proliferation fied. We interrogated this pathway here, because muscle
of the satellite cell population as opposed to differentiation. deconditioning and relative disuse can occur in the COPD
This would impair the regenerative capacity of COPD muscle, population. However, it appears that, in the patient with stable
and potentially contribute to the loss of muscle mass. COPD, p50 engagement does not contribute to muscle atrophy.
The NF-kB signaling pathway is known to mediate muscle Whether there is an incremental effect, with p50 recruitment
atrophy in rodent models of muscle wasting associated with during acute exacerbations of COPD in a comparable patient
cachexia, systemic inflammation, denervation, and unloading (7, population, remains to be determined.
8, 20, 23, 50) through the combined effects of stimulated Phosphorylation and activation of AKT is well known to
proteolysis and impaired muscle growth. Of the five known induce skeletal muscle hypertrophy (6, 25). AKT activation
transcription factors (p65, Rel B, C-rel, p52, and p50), p65 is the results in the recruitment and subsequent phosphorylation of
primary subunit known to regulate muscle biology. As with all multiple direct and indirect downstream targets, including
NF-kB transcription factors, p65 heterodimers are held in the mammalian target of rapamycin, p70S6 kinase, PHAS-1, and/
cytosol in an inactive complex with IkB (24, 51, 52). p65 or GSK3b, all of which contribute to the development of muscle
activation occurs via phosphorylation of IkBa, release, and hypertrophy, at least in part, via the induction of protein
translocation to the nucleus. Phosphorylation of NF-kB(p65) in synthesis. It has also been recently appreciated that activation
the nucleus enhances its transcriptional activity. It is there that of AKT signaling suppresses the expression of atrogin-1 and
the p65/p50 heterodimers bind the MuRF1 promotor, inducing MuRF1 via phosphorylation of the Forkhead O transcription
MuRF1 transcription and proteasomal mediated muscle degra- factors, and thereby actively suppresses muscle proteolysis (25,
dation (23). p65 is the sole subunit currently shown to regulate 26). Conversely, down-regulation of AKT signaling occurs in
myogenesis (24, 53). muscle atrophied by denervation, disuse, and glucocorticoid
There are reports suggesting that up-regulation of NF-kB administration, demonstrating reciprocal communication be-
signaling is involved in the development of muscle atrophy tween pathways mediating muscle atrophy and hypertrophy
associated with COPD. Agusti and colleagues (50) demon- (8). The amount of material available to us from the muscle
strated NF-kB(p65) activation in pooled muscle protein lysates biopsy limited the extent to which we were able to interrogate
from patients with COPD with low BMI, compared with the AKT signaling network, and we focused our assessment on
patients with COPD with normal to high BMI. Inflammatory AKT, GSK3b, and p70S6 kinase. Surprisingly, despite the
cytokines, such as TNF-a, induce muscle atrophy by engaging atrophy seen in the COPD population, we were unable to
NF-kB(p65) signaling, which results in increased proteasomal- demonstrate down-regulation of AKT signaling in our cohort of
mediated protein degradation and inhibition of muscle regen- patients with COPD.
erative pathways via the induction of oxidative stress, and In contrast, Doucet and colleagues (37) demonstrated an
decreased MyoD expression (20, 22). Low-grade systemic in- increase in protein levels of phosphorylated AKT, GSK3b, and
flammation (elevated circulating plasma levels of TNF-a, and p70S6 kinase in patients with COPD, and hypothesized a com-
IL-1 and -6) has been described in patients with COPD (54, 55). pensatory mechanism of muscle mass recovery accounting for
Despite these reports of up-regulation of systemic inflammation this activation (37). The discrepant findings between our study
in patients with COPD at both a resting state and during and that by Doucet and colleagues may be attributable to
acute exacerbations, and in contrast to Agustis findings, we differences employed in the normalization of data, yet the
were unable to demonstrate engagement of IkBa or NF- critical observation is that neither study found AKT signaling
kB(p65) in the vastus lateralis muscles of our patients with to be down-regulated in the patients with COPD. This was
COPD. The absence of engagement of NF-kB(p65) signaling in unexpected and in contrast to data generated in rodent models
our patients is supported by normal expression levels of MuRF1 of muscle atrophy. This finding suggests the possibility that
and MyoD, both downstream of p65. In contrast to Agustis a compensatory response is in place in the muscle of patients
patient cohort, our patient population had a normal BMI, which with COPD that attempts to counteract the atrophic stimuli.
may have contributed to the difference in findings between our There is certainly precedent for this phenomenon. Attempts at
studies. muscle recovery after muscle atrophy are described in other
470 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 42 2010

models (59), but this recovery response is temporally limited, chronic obstructive pulmonary disease. Respir Med 2000;94:859
and can be exhausted. 867.
3. Decramer M, Gosselink R, Troosters T, Verschueren M, Evers G.
Activation of autophagy has recently been shown to con-
Muscle weakness is related to utilization of health care resources in
tribute to the development of muscle atrophy. LC3 and beclin-1, COPD patients. Eur Respir J 1997;10:417423.
both known components of autophagy signaling, have been 4. Marquis K, Debigare R, Lacasse Y, LeBlanc P, Jobin J, Carrier G,
shown to increase in rodent models of denervation and unloading- Maltais F. Midthigh muscle cross-sectional area is a better predictor
induced muscle atrophy (18). In C2C12 myotubes in vitro, of mortality than body mass index in patients with chronic obstructive
autophagy has been demonstrated to play as important a role pulmonary disease. Am J Respir Crit Care Med 2002;166:809813.
as ubiquitinproteasomemediated muscle proteolysis (19). In 5. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss is a re-
addition, AKT signaling has been shown to be reciprocally versible factor in the prognosis of chronic obstructive pulmonary
disease. Am J Respir Crit Care Med 1998;157:17911797.
linked to autophagy, as it has been to ubiquitin-mediated deg- 6. Glass DJ. Skeletal muscle hypertrophy and atrophy signaling pathways.
radation (60, 61). Decreased AKT signaling results in relative Int J Biochem Cell Biol 2005;37:19741984.
dephosphorylation and nuclear translocation of Forkhead O 7. Hansen MJ, Gualano RC, Bozinovski S, Vlahos R, Anderson GP.
transcription factors, which, in turn, results in the increased Therapeutic prospects to treat skeletal muscle wasting in
expression of multiple autophagy-related genes and the in- COPD (chronic obstructive lung disease). Pharmacol Ther 2006;109:
duction of autophagy. Regardless of the mounting evidence 162172.
detailing the importance of autophagy in skeletal muscle 8. Kandarian SC, Jackman RW. Intracellular signaling during skeletal
muscle atrophy. Muscle Nerve 2006;33:155165.
atrophy, we did not note an increase in beclin-1 or LC3
9. Medina R, Wing SS, Goldberg AL. Increase in levels of polyubiquitin
transcript levels in our COPD population. and proteasome mRNA in skeletal muscle during starvation and
Although our study carefully describes molecular changes denervation atrophy. Biochem J 1995;307:631637.
seen in muscle atrophy associated with stable COPD compared 10. Price SR, Bailey JL, Wang X, Jurkovitz C, England BK, Ding X, Phillips
with that in control subjects, there are some limitations inherent LS, Mitch WE. Muscle wasting in insulinopenic rats results from
in clinical molecular studies. First, we were unable to identically activation of the ATP-dependent, ubiquitinproteasome proteolytic
match our control and COPD populations, with more females in pathway by a mechanism including gene transcription. J Clin Invest
1996;98:17031708.
the control group. However, to date, we are unaware of data
11. Attaix D, Ventadour S, Codran A, Bechet D, Taillandier D, Combaret
that support pathways inducing muscle atrophy that are differ- L. The ubiquitinproteasome system and skeletal muscle wasting.
entially regulated in males and females. Unlike rodent studies, Essays Biochem 2005;41:173186.
limited clinical material requires a selective, rather than ex- 12. Lecker SH, Goldberg AL, Mitch WE. Protein degradation by the
haustive, evaluation of molecular pathways, and the total ubiquitinproteasome pathway in normal and disease states. J Am
number of participants was small. There is a risk that the study Soc Nephrol 2006;17:18071819.
is underpowered as a result, and further work is required to 13. Batt J, Bain J, Goncalves J, Michalski B, Plant P, Fahnestock M,
complement our study. Finally, a dearth of reliable antibodies Woodgett J. Differential gene expression profiling of short and long
term denervated muscle. FASEB J 2006;20:115117.
for specific targets of interest, combined with limited clinical 14. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA,
material, restricts some of the data to transcriptional mRNA Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, et al. Identifica-
expression only, but we believe that this is reflective of total tion of ubiquitin ligases required for skeletal muscle atrophy. Science
cellular protein levels for the mediators that we assessed. 2001;294:17041708.
Thus, we have demonstrated, in a well-characterized cohort 15. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1,
of individuals with severe COPD and decreased skeletal muscle a muscle-specific F-box protein highly expressed during muscle
mass, the up-regulation of mediators of protein degradation atrophy. Proc Natl Acad Sci USA 2001;98:1444014445.
16. Koncarevic A, Jackman RW, Kandarian SC. The ubiquitinprotein
(atrogin-1 and Nedd4) and of the myogenic inhibitory factor,
ligase Nedd4 targets Notch1 in skeletal muscle and distinguishes the
myostatin, in the vastus lateralis muscle. These findings imply subset of atrophies caused by reduced muscle tension. FASEB J 2007;
that the muscle atrophy associated with COPD results from 21:427437.
both an inhibition of the reparative capacity of muscle and 17. Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price
increased ubiquitin-mediated proteolysis, and that there is SR, Mitch WE, Goldberg AL. Multiple types of skeletal muscle
specificity in the ubiquitin ligases recruited to the etiology of atrophy involve a common program of changes in gene expression.
muscle atrophy in humans. Whether the changes in the biology FASEB J 2004;18:3951.
18. OLeary MF, Hood DA. Denervation-induced oxidative stress and
of the muscle in the population of patients with COPD results
autophagy signaling in muscle. Autophagy 2009;5:230231.
from the lung disease itself, or the combination of a chronic 19. Zhao J, Brault JJ, Schild A, Goldberg AL. Coordinate activation of
disease accompanied by possible altered nutritional status and/ autophagy and the proteasome pathway by FoxO Transcription
or diminished activity, is not clear. Regardless, characterizing Factor. Autophagy 2008;4:378380.
the biological networks activated in the atrophied muscle of 20. Langen RC, Van Der Velden JL, Schols AM, Kelders MC, Wouters EF,
humans with end-stage COPD is necessary to allow the de- Janssen-Heininger YM. Tumor necrosis factor-alpha inhibits myo-
velopment of therapeutic interventions to counteract muscle genic differentiation through MyoD protein destabilization. FASEB J
atrophy and to improve patient function, quality of life, and, 2004;18:227237.
21. Spate U, Schulze PC. Proinflammatory cytokines and skeletal muscle.
potentially, survival.
Curr Opin Clin Nutr Metab Care 2004;7:265269.
Conflict of Interest Statement: None of the authors has a financial relationship 22. Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin AS Jr. NF-
with a commercial entity that has an interest in the subject of this manuscript. kappaB-induced loss of MyoD messenger RNA: possible role in
muscle decay and cachexia. Science 2000;289:23632366.
Acknowledgments: The authors thank Forough Farrokhyar for her assistance with 23. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, Lidov HG,
the statistical analysis. Hasselgren PO, Frontera WR, Lee J, Glass DJ, et al. IKKbeta/NF-
kappaB activation causes severe muscle wasting in mice. Cell 2004;
References 119:285298.
1. Gosselink R, Troosters T, Decramer M. Peripheral muscle weakness 24. Mourkioti F, Rosenthal N. NF-kappaB signaling in skeletal muscle:
contributes to exercise limitation in COPD. Am J Respir Crit Care prospects for intervention in muscle diseases. J Mol Med 2008;86:747
Med 1996;153:976980. 759.
2. Mostert R, Goris A, Weling-Scheepers C, Wouters EF, Schols AM. 25. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R,
Tissue depletion and health related quality of life in patients with Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, et al. Akt/mTOR
Plant, Brooks, Faughnan, et al.: Markers of Muscle Atrophy in COPD 471

pathway is a crucial regulator of skeletal muscle hypertrophy and can 44. Tintignac LA, Lagirand J, Batonnet S, Sirri V, Leibovitch MP, Leibovitch
prevent muscle atrophy in vivo. Nat Cell Biol 2001;3:10141019. SA. Degradation of MyoD mediated by the SCF (MAFbx) ubiquitin
26. Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, ligase. J Biol Chem 2005;280:28472856.
Gonzalez M, Yancopoulos GD, Glass DJ. The IGF-1/PI3K/Akt path- 45. Plant PJ, Lafont F, Lecat S, Verkade P, Simons K, Rotin D. Apical
way prevents expression of muscle atrophy-induced ubiquitin ligases by membrane targeting of Nedd4 is mediated by an association of its C2
inhibiting FOXO transcription factors. Mol Cell 2004;14:395403. domain with annexin XIIIb. J Cell Biol 2000;149:14731484.
27. Walsh FS, Celeste AJ. Myostatin: a modulator of skeletal-muscle stem 46. Anan T, Nagata Y, Koga H, Honda Y, Yabuki N, Miyamoto C, Kuwano
cells. Biochem Soc Trans 2005;33:15131517. A, Matsuda I, Endo F, Saya H, et al. Human ubiquitinprotein ligase
28. ODonnell DE, Aaron S, Bourbeau J, Hernandez P, Marciniuk D, Nedd4: expression, subcellular localization and selective interaction
Balter M, Ford G, Gervais A, Goldstein R, Hodder R, et al. State of with ubiquitin-conjugating enzymes. Genes Cells 1998;3:751763.
the art compendium: Canadian Thoracic Society recommendations 47. Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle
for the management of chronic obstructive pulmonary disease. Can regeneration. Physiol Rev 2004;84:209238.
Respir J 2004;11(Suppl B):7B59B. 48. Hawke TJ, Garry DJ. Myogenic satellite cells: physiology to molecular
29. Miller MR, Hankinson J, Brusasco V, Burgo F, Casaburi R, Coates A, biology. J Appl Physiol 2001;91:534551.
Crapo R, Enright P, van der Grinten CP, Gustafsson P, et al.; ATS/ 49. Gonzalez-Cadavid NF, Taylor WE, Yarasheski K, Sinha-Hikim I, Ma K,
ERS Task Force. Standardisation of spirometry. Eur Respir J 2005;26: Ezzat S, Shen R, Lalani R, Asa S, Mamita M, et al. Organization of
319338. the human myostatin gene and expression in healthy men and HIV-
30. Brooks D, Solway S, Gibbons WJ. ATS statement on six-minute walk infected men with muscle wasting. Proc Natl Acad Sci USA 1998;95:
test. Am J Respir Crit Care Med 2003;167:1287. 1493814943.
31. Amercian Thoracic Society. ATS statement: guidelines for the six- 50. Agusti A, Morla M, Sauleda J, Saus C, Busquets X. NF-kappaB
minute walk test. Am J Respir Crit Care Med 2002;166:111117. activation and iNOS upregulation in skeletal muscle of patients with
32. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M, Mendez COPD and low body weight. Thorax 2004;59:483487.
RA, Pinto Plata, V, Cabral HJ. The body-mass index, airflow 51. Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat
obstruction, dyspnea, and exercise capacity index in chronic obstruc- Rev Mol Cell Biol 2004;5:392401.
tive pulmonary disease. N Engl J Med 2004;350:10051012. 52. Yamamoto Y, Gaynor RB. Ikappa B kinases: key regulators of the NF-
33. Bernard S, LeBlanc P, Whittom F, Carrier G, Jobin J, Belleau R, Maltais kappaB pathway. Trends Biochem Sci 2004;29:7279.
F. Peripheral muscle weakness in patients with chronic obstructive 53. Acharyya S, Villalta SA, Bakkar N, Bupha-Intr T, Janssen PM,
pulmonary disease. Am J Respir Crit Care Med 1998;158:629634. Carathers M, Li ZW, Beg AA, Ghosh S, Sahenk Z, et al. Interplay
34. Hagberg M. Muscular endurance and surface electromyogram in iso- of IKK/NF-kappaB signaling in macrophages and myofibers promotes
metric and dynamic exercise. J Appl Physiol 1981;51:17. muscle degeneration in Duchenne muscular dystrophy. J Clin Invest
35. Todd G, Gorman RB, Gandevia SC. Measurement and reproducibility 2007;117:889901.
of strength and voluntary activation of lower-limb muscles. Muscle 54. Gan WQ, Man SF, Senthilselvan A, Sin DD. Association between
Nerve 2004;29:834842. chronic obstructive pulmonary disease and systemic inflammation:
36. Bourgeois JM, Tarnopolsky MA. Pathology of skeletal muscle in a systematic review and a meta-analysis. Thorax 2004;59:574580.
mitochondrial disorders. Mitochondrion 2004;4:441452. 55. Schols AM, Buurman WA, Staal van den Brekel AJ, Dentener MA,
37. Doucet M, Russell AP, Leger B, Debigare R, Joanisse DR, Caron MA, Wouters EF. Evidence for a relation between metabolic derange-
LeBlanc P, Maltais F. Muscle atrophy and hypertrophy signaling in ments and increased levels of inflammatory mediators in a subgroup
patients with chronic obstructive pulmonary disease. Am J Respir Crit of patients with chronic obstructive pulmonary disease. Thorax 1996;
Care Med 2007;176:261269. 51:819824.
38. Pickart CM. Mechanisms underlying ubiquitination. Annu Rev Biochem 56. Barreiro E, Schols AM, Polkey MI, Galdiz JB, Gosker HR, Swallow EB,
2001;70:503533. Coronell C, Gea J.; ENIGMA in COPD Project. Cytokine profile in
39. Plant PJ, Correa J, Goldenberg N, Bain J, Batt J. The inositol quadriceps muscles of patients with severe chronic obstructive
phosphatase MTMR4 is a novel target of the ubiquitin ligase Nedd4. pulmonary disease. Thorax 2008;63:100107.
Biochem J 2009;419:5763. 57. Crul T, Spruit MA, Gayan-Ramirez G, Quarck R, Gosselink R,
40. Ottenheijm CA, Heunks LM, Li YP, Jin B, Minnaard R, van Hees HW, Troosters T, Pitta F, Decramer M. Markers of inflammation and
Dekhuijzen PN. Activation of the ubiquitinproteasome pathway in disuse in vastus lateralis of chronic obstructive pulmonary disease
the diaphragm in chronic obstructive pulmonary disease. Am J Respir patients. Eur J Clin Invest 2007;37:897904.
Crit Care Med 2006;174:9971002. 58. Hunter RB, Kandarian SC. Disruption of either the NFkb1 or the Bcl3
41. Stevenson EJ, Giresi PG, Koncarevic A, Kandarian SC. Global analysis gene inhibits skeletal muscle atrophy. J Clin Invest 2004;114:15041511.
of gene expression patterns during disuse atrophy in rat skeletal 59. Irintchev A, Draguhn A, Wernig A. Reinnervation and recovery of
muscle. J Physiol 2003;551:3348. mouse soleus muscle after long-term denervation. Neuroscience 1990;
42. Witt SH, Granzier H, Witt CC, Labeit S. MURF-1 and MURF-2 target 39:231243.
a specific subset of myofibrillar proteins redundantly: towards un- 60. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH,
derstanding MURF-dependent muscle ubiquitination. J Mol Biol Goldberg AL. FoxO3 coordinately activates protein degradation by
2005;350:713722. the autophagic/lysosomal and proteasomal pathways in atrophying
43. Kedar V, McDonough H, Arya R, Li HH, Rockman HA, Patterson C. muscle cells. Cell Metab 2007;6:472483.
Muscle-specific RING Finger 1 is a bona fide ubiquitin ligase that 61. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del PP,
degrades cardiac troponin I. Proc Natl Acad Sci USA 2004;101:18135 Burden SJ, Di LR, Sandri C, Zhao J, et al. FoxO3 controls autophagy
18140. in skeletal muscle in vivo. Cell Metab 2007;6:458471.

Das könnte Ihnen auch gefallen